Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
J Pediatr Endocrinol Metab ; 35(10): 1306-1308, 2022 Oct 26.
Artigo em Inglês | MEDLINE | ID: mdl-35932482

RESUMO

OBJECTIVES: Congenital hypogonadotropic hypogonadism combined with anosmia or hyposmia is considered Kallmann syndrome (KS). It is often accompanied by bone defects. CASE PRESENTATION: Here, we report a girl and her mother with KS caused by a novel mutation in the fibroblast growth factor receptor 1 gene (FGFR1). Interestingly, the daughter presented syndactyly and oligodactyly of the feet. CONCLUSIONS: The presence of bone malformations in a KS patient should direct the geneticist towards a search for specific mutations in FGFR1. Our finding contributes to enrich the spectrum of FGFR1 mutations in patients with KS.


Assuntos
Hipogonadismo , Síndrome de Kallmann , Feminino , Humanos , Hipogonadismo/genética , Síndrome de Kallmann/genética , Mães , Mutação , Núcleo Familiar , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/genética , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/metabolismo
2.
Clin Genet ; 97(1): 114-124, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31355911

RESUMO

Hirschsprung disease (HSCR) is a rare congenital disorder caused by an incorrect enteric nervous system development due to a failure in migration, proliferation, differentiation and/or survival of enteric neural crest cells. HSCR is a complex genetic disease, where alterations at different molecular levels are required for the manifestation of the disease. In addition, a wide spectrum of mutations affecting many different genes cause HSCR, although the occurrence and severity of HSCR from many cases still remain unexplained. This review summarizes the current knowledge about molecular genetic basis of HSCR.


Assuntos
Doença de Hirschsprung/genética , Doenças do Sistema Nervoso/genética , Sistema Nervoso/patologia , Proteínas Proto-Oncogênicas c-ret/genética , Doença de Hirschsprung/patologia , Humanos , Mutação/genética , Sistema Nervoso/metabolismo , Doenças do Sistema Nervoso/patologia , Crista Neural/crescimento & desenvolvimento , Crista Neural/patologia , Transdução de Sinais/genética
3.
Neuromuscul Disord ; 28(3): 208-215, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29433793

RESUMO

Spinal muscular atrophy (SMA) is a neuromuscular disorder caused by loss or mutations in SMN1. According to age of onset, achieved motor abilities, and life span, SMA patients are classified into type I (never sit), II (never walk unaided) or III (achieve independent walking abilities). SMN2, the highly homologous copy of SMN1, is considered the most important phenotypic modifier of the disease. Determination of SMN2 copy number is essential to establish careful genotype-phenotype correlations, predict disease evolution, and to stratify patients for clinical trials. We have determined SMN2 copy numbers in 625 unrelated Spanish SMA patients with loss or mutation of both copies of SMN1 and a clear assignation of the SMA type by clinical criteria. Furthermore, we compiled data from relevant worldwide reports that link SMN2 copy number with SMA severity published from 1999 to date (2834 patients with different ethnic and geographic backgrounds). Altogether, we have assembled a database with a total of 3459 patients to delineate more universal prognostic rules regarding the influence of SMN2 copy number on SMA phenotype. This issue is crucial in the present scenario of therapeutic advances with the perspective of SMA neonatal screening and early diagnosis to initiate treatments.


Assuntos
Variações do Número de Cópias de DNA , Estudos de Associação Genética , Atrofia Muscular Espinal/genética , Bases de Dados Genéticas , Feminino , Dosagem de Genes , Predisposição Genética para Doença , Genótipo , Humanos , Masculino , Mutação , Fenótipo , Prognóstico , Espanha , Proteína 2 de Sobrevivência do Neurônio Motor/genética
5.
Pediatr Dev Pathol ; 20(1): 28-37, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28276298

RESUMO

Objectives Hirschsprung disease (HSCR) has a wide range of severity. There are nonsevere forms treated conservatively until surgery and severe forms that require an early stoma and prolonged hospitalization. Our objective was to establish a clinical scoring system to predict the severity of HSCR and to evaluate the possible existence of a clinical-genetic correlation. Methods We carried out a retrospective observational study including all HSCR cases treated in our hospital. The sample was divided into severe and nonsevere disease according to the number of surgical procedures, hospitalization time, and episodes of enterocolitis. The proposed score was applied at diagnosis, and the sensitivity, specificity, and optimal cut-point were determined. We conducted a prospective molecular study of RET, EDNRB, and EDN3 on all patients, as well as SOX10 in Waardenburg Syndrome type 4 forms. Results Among the 42 patients treated between 1983 and 2013, 15 met the severe disease criteria. This group had a higher mean score (13.15 ± 2.36) than the nonsevere group (8.15 ± 2.13; p < 0.001). A score ≥11 had a sensitivity of 87% and a specificity of 81% in detecting the severe cases. Causative mutations were identified in 12 patients, 8 of them in the severe group ( p = 0.015). Most of these mutations (75%) were located in the RET proto-oncogene. Conclusion The proposed scoring system enables the early selection of patients with severe behavior of HSCR. A value ≥11 showed good sensitivity and specificity for this purpose. Causative mutations were identified in more than 50% of patients who met the criteria for severe disease.


Assuntos
Estudos de Associação Genética , Marcadores Genéticos , Doença de Hirschsprung/diagnóstico , Mutação , Índice de Gravidade de Doença , Adolescente , Criança , Pré-Escolar , Feminino , Seguimentos , Doença de Hirschsprung/genética , Humanos , Lactente , Recém-Nascido , Modelos Logísticos , Masculino , Proto-Oncogene Mas , Estudos Retrospectivos , Sensibilidade e Especificidade
6.
Hum Mol Genet ; 25(23): 5265-5275, 2016 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-27702942

RESUMO

Hirschsprung disease (HSCR) is the most common cause of neonatal intestinal obstruction. It is characterized by the absence of ganglia in the nerve plexuses of the lower gastrointestinal tract. So far, three common disease-susceptibility variants at the RET, SEMA3 and NRG1 loci have been detected through genome-wide association studies (GWAS) in Europeans and Asians to understand its genetic etiologies. Here we present a trans-ethnic meta-analysis of 507 HSCR cases and 1191 controls, combining all published GWAS results on HSCR to fine-map these loci and narrow down the putatively causal variants to 99% credible sets. We also demonstrate that the effects of RET and NRG1 are universal across European and Asian ancestries. In contrast, we detected a European-specific association of a low-frequency variant, rs80227144, in SEMA3 [odds ratio (OR) = 5.2, P = 4.7 × 10-10]. Conditional analyses on the lead SNPs revealed a secondary association signal, corresponding to an Asian-specific, low-frequency missense variant encoding RET p.Asp489Asn (rs9282834, conditional OR = 20.3, conditional P = 4.1 × 10-14). When in trans with the RET intron 1 enhancer risk allele, rs9282834 increases the risk of HSCR from 1.1 to 26.7. Overall, our study provides further insights into the genetic architecture of HSCR and has profound implications for future study designs.


Assuntos
Predisposição Genética para Doença , Doença de Hirschsprung/genética , Neuregulina-1/genética , Proteínas Proto-Oncogênicas c-ret/genética , Semaforina-3A/genética , Alelos , Povo Asiático/genética , Etnicidade/genética , Feminino , Estudo de Associação Genômica Ampla , Genótipo , Doença de Hirschsprung/patologia , Humanos , Íntrons/genética , Masculino , Polimorfismo de Nucleotídeo Único , População Branca/genética
7.
Clin Case Rep ; 4(9): 879-84, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27648268

RESUMO

Despite co-segregation of two different genetic neurological disorders within a family is rare, clinicians should take into consideration this possibility in patients presenting with unusual complex phenotypes or with unexpected electrophysiological findings. Here, we report a Spanish 11-month-old patient with spinal muscular atrophy type 2 and Charcot-Marie-Tooth 1A.

9.
Am J Med Genet A ; 170A(5): 1268-73, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26762557

RESUMO

Monosomy 10p is a rare chromosomal disorder with a prevalence <1/1,000,000, in which a terminal or interstitial distal region of chromosome 10 is deleted resulting in a variable phenotype depending on the size of the deletion. Two main phenotypes have been defined depending on the location of the deletion: HDR syndrome (Hypoparathyroidism, sensorineural Deafness, and Renal disease), and DGS2 (DiGeorge syndrome type 2). The vast majority of cases reported so far have resulted from de novo events. Here, we present the first familial presentation of this contiguous gene deletion syndrome, affecting two family members in different generations: a child and his maternal uncle. In both cases, the deletion was due to a malsegregation of a maternal balanced rearrangement, ins(16;10)(q22;p13p15.2). The identification and characterization of this rearrangement was possible using a combination of different genetic analyses such as karyotype, MLPA, FISH, and array CGH. We underline the importance of the present results in terms of genetic and reproductive counseling for the carriers of the balanced rearrangement within the family, and demonstrate again the utility of expanding the genetic studies to the relatives of the affected patients.


Assuntos
Transtornos Cromossômicos/genética , Cromossomos Humanos Par 10/genética , Síndrome de DiGeorge/genética , Perda Auditiva Neurossensorial/genética , Hipoparatireoidismo/genética , Nefrose/genética , Adulto , Criança , Deleção Cromossômica , Transtornos Cromossômicos/fisiopatologia , Hibridização Genômica Comparativa , Deficiências do Desenvolvimento/genética , Deficiências do Desenvolvimento/fisiopatologia , Síndrome de DiGeorge/fisiopatologia , Deleção de Genes , Perda Auditiva Neurossensorial/fisiopatologia , Humanos , Hipoparatireoidismo/fisiopatologia , Hibridização in Situ Fluorescente , Cariotipagem , Masculino , Nefrose/fisiopatologia
10.
BMC Med Genomics ; 8: 83, 2015 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-26690675

RESUMO

BACKGROUND: The molecular mechanisms leading to sporadic medullary thyroid carcinoma (sMTC) and juvenile papillary thyroid carcinoma (PTC), two rare tumours of the thyroid gland, remain poorly understood. Genetic studies on thyroid carcinomas have been conducted, although just a few loci have been systematically associated. Given the difficulties to obtain single-loci associations, this work expands its scope to the study of epistatic interactions that could help to understand the genetic architecture of complex diseases and explain new heritable components of genetic risk. METHODS: We carried out the first screening for epistasis by Multifactor-Dimensionality Reduction (MDR) in genome-wide association study (GWAS) on sMTC and juvenile PTC, to identify the potential simultaneous involvement of pairs of variants in the disease. RESULTS: We have identified two significant epistatic gene interactions in sMTC (CHFR-AC016582.2 and C8orf37-RNU1-55P) and three in juvenile PTC (RP11-648k4.2-DIO1, RP11-648k4.2-DMGDH and RP11-648k4.2-LOXL1). Interestingly, each interacting gene pair included a non-coding RNA, providing thus support to the relevance that these elements are increasingly gaining to explain carcinoma development and progression. CONCLUSIONS: Overall, this study contributes to the understanding of the genetic basis of thyroid carcinoma susceptibility in two different case scenarios such as sMTC and juvenile PTC.


Assuntos
Carcinoma Neuroendócrino/genética , Carcinoma/genética , Epistasia Genética , Estudo de Associação Genômica Ampla , Neoplasias da Glândula Tireoide/genética , Adolescente , Carcinoma Papilar , Estudos de Casos e Controles , Criança , Pré-Escolar , Feminino , Predisposição Genética para Doença/genética , Humanos , Masculino , Polimorfismo de Nucleotídeo Único , Câncer Papilífero da Tireoide , Adulto Jovem
11.
Biomed Res Int ; 2015: 965839, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26713318

RESUMO

Fragile X syndrome (FXS) accounts for about one-half of cases of X-linked intellectual disability and is the most common monogenic cause of mental impairment. Reproductive options for the FXS carriers include preimplantation genetic diagnosis (PGD). However, this strategy is considered by some centers as wasteful owing to the high prevalence of premature ovarian failure in FXS carriers and the difficulties in genetic diagnosis of the embryos. Here we present the results of our PGD Program applied to FXS, at the Department of Genetics, Reproduction and Fetal Medicine of the University Hospital Virgen del Rocío in Seville. A total of 11 couples have participated in our PGD Program for FXS since 2010. Overall, 15 cycles were performed, providing a total of 43 embryos. The overall percentage of transfers per cycle was 46.67% and the live birth rate per cycle was 13.33%. As expected, these percentages are considerably lower than the ones obtained in PGD for other pathologies. Our program resulted in the birth of 3 unaffected babies of FXS for 2 of the 11 couples (18.2%) supporting that, despite the important drawbacks of PGD for FXS, efforts should be devoted in offering this reproductive option to the affected families.


Assuntos
Síndrome do Cromossomo X Frágil/diagnóstico , Testes Genéticos , Diagnóstico Pré-Implantação , Insuficiência Ovariana Primária/diagnóstico , Adulto , Coeficiente de Natalidade , Transferência Embrionária , Feminino , Síndrome do Cromossomo X Frágil/genética , Síndrome do Cromossomo X Frágil/patologia , Heterozigoto , Hospitais Universitários , Humanos , Gravidez , Insuficiência Ovariana Primária/genética , Insuficiência Ovariana Primária/patologia , Espanha
12.
Sci Rep ; 5: 16473, 2015 Nov 12.
Artigo em Inglês | MEDLINE | ID: mdl-26559152

RESUMO

Hirschsprung disease (HSCR; OMIM 142623) is a developmental disorder characterized by aganglionosis along variable lengths of the distal gastrointestinal tract, which results in intestinal obstruction. Interactions among known HSCR genes and/or unknown disease susceptibility loci lead to variable severity of phenotype. Neither linkage nor genome-wide association studies have efficiently contributed to completely dissect the genetic pathways underlying this complex genetic disorder. We have performed whole exome sequencing of 16 HSCR patients from 8 unrelated families with SOLID platform. Variants shared by affected relatives were validated by Sanger sequencing. We searched for genes recurrently mutated across families. Only variations in the FAT3 gene were significantly enriched in five families. Within-family analysis identified compound heterozygotes for AHNAK and several genes (N = 23) with heterozygous variants that co-segregated with the phenotype. Network and pathway analyses facilitated the discovery of polygenic inheritance involving FAT3, HSCR known genes and their gene partners. Altogether, our approach has facilitated the detection of more than one damaging variant in biologically plausible genes that could jointly contribute to the phenotype. Our data may contribute to the understanding of the complex interactions that occur during enteric nervous system development and the etiopathology of familial HSCR.


Assuntos
Exoma , Heterogeneidade Genética , Sequenciamento de Nucleotídeos em Larga Escala , Doença de Hirschsprung/genética , Alelos , Caderinas/genética , Fator de Crescimento Epidérmico/genética , Família , Feminino , Estudo de Associação Genômica Ampla , Doença de Hirschsprung/diagnóstico , Humanos , Padrões de Herança , Masculino , Mutação , Linhagem , Fenótipo , Polimorfismo de Nucleotídeo Único
13.
Biomed Res Int ; 2015: 406096, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26258137

RESUMO

Hemophilia A and B are the most common hereditary hemorrhagic disorders, with an X-linked mode of inheritance. Reproductive options for the families affected with hemophilia, aiming at the prevention of the birth of children with severe coagulation disorders, include preimplantation genetic diagnosis (PGD). Here we present the results of our PGD Program applied to hemophilia, at the Department of Genetics, Reproduction and Fetal Medicine of the University Hospital Virgen del Rocío in Seville. A total of 34 couples have been included in our program since 2005 (30 for hemophilia A and 4 for hemophilia B). Overall, 60 cycles were performed, providing a total of 508 embryos. The overall percentage of transfers per cycle was 81.7% and the live birth rate per cycle ranged from 10.3 to 24.1% depending on the methodological approach applied. Although PGD for hemophilia can be focused on gender selection of female embryos, our results demonstrate that methodological approaches that allow the diagnosis of the hemophilia status of every embryo have notorious advantages. Our PGD Program resulted in the birth of 12 healthy babies for 10 out of the 34 couples (29.4%), constituting a relevant achievement for the Spanish Public Health System within the field of haematological disorders.


Assuntos
Testes Genéticos/métodos , Hemofilia A/diagnóstico , Hemofilia A/genética , Hemofilia B/diagnóstico , Hemofilia B/genética , Hospitais Universitários , Diagnóstico Pré-Implantação/métodos , Adulto , Embrião de Mamíferos/fisiologia , Feminino , Humanos , Hibridização in Situ Fluorescente , Masculino , Repetições de Microssatélites/genética , Reação em Cadeia da Polimerase , Gravidez , Espanha
14.
Am J Hum Genet ; 96(4): 581-96, 2015 Apr 02.
Artigo em Inglês | MEDLINE | ID: mdl-25839327

RESUMO

Innervation of the gut is segmentally lost in Hirschsprung disease (HSCR), a consequence of cell-autonomous and non-autonomous defects in enteric neuronal cell differentiation, proliferation, migration, or survival. Rare, high-penetrance coding variants and common, low-penetrance non-coding variants in 13 genes are known to underlie HSCR risk, with the most frequent variants in the ret proto-oncogene (RET). We used a genome-wide association (220 trios) and replication (429 trios) study to reveal a second non-coding variant distal to RET and a non-coding allele on chromosome 7 within the class 3 Semaphorin gene cluster. Analysis in Ret wild-type and Ret-null mice demonstrates specific expression of Sema3a, Sema3c, and Sema3d in the enteric nervous system (ENS). In zebrafish embryos, sema3 knockdowns show reduction of migratory ENS precursors with complete ablation under conjoint ret loss of function. Seven candidate receptors of Sema3 proteins are also expressed within the mouse ENS and their expression is also lost in the ENS of Ret-null embryos. Sequencing of SEMA3A, SEMA3C, and SEMA3D in 254 HSCR-affected subjects followed by in silico protein structure modeling and functional analyses identified five disease-associated alleles with loss-of-function defects in semaphorin dimerization and binding to their cognate neuropilin and plexin receptors. Thus, semaphorin 3C/3D signaling is an evolutionarily conserved regulator of ENS development whose dys-regulation is a cause of enteric aganglionosis.


Assuntos
Epistasia Genética/genética , Predisposição Genética para Doença/genética , Variação Genética , Doença de Hirschsprung/genética , Proteínas Proto-Oncogênicas c-ret/genética , Semaforinas/genética , Animais , Sequência de Bases , Estudo de Associação Genômica Ampla , Camundongos , Dados de Sequência Molecular , Semaforinas/deficiência , Semaforinas/metabolismo , Análise de Sequência de DNA
15.
Am J Med Genet A ; 164A(2): 542-7, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24311220

RESUMO

Shah-Waardenburg syndrome or Waardenburg syndrome type 4 (WS4) is a neurocristopathy characterized by the association of deafness, depigmentation and Hirschsprung disease. Three disease-causing genes have been identified so far for WS4: EDNRB, EDN3, and SOX10. SOX10 mutations, found in 45-55% of WS4 patients, are inherited in autosomal dominant way. In addition, mutations in SOX10 are also responsible for an extended syndrome involving peripheral and central neurological phenotypes, referred to as PCWH (peripheral demyelinating neuropathy, central dysmyelinating leucodystrophy, Waardenburg syndrome, Hirschsprung disease). Such mutations are mostly private, and a high intra- and inter-familial variability exists. In this report, we present a patient with WS4 and a second with PCWH due to SOX10 mutations supporting again the genetic and phenotypic heterogeneity of these syndromes. Interestingly, the WS4 family carries an insertion of 19 nucleotides in exon 5 of SOX10, which results in distinct phenotypes along three different generations: hypopigmentation in the maternal grandmother, hearing loss in the mother, and WS4 in the proband. Since mosaicism cannot explain the three different related-WS features observed in this family, we propose as the most plausible explanation the existence of additional molecular events, acting in an additive or multiplicative fashion, in genes or regulatory regions unidentified so far. On the other hand, the PCWH case was due to a de novo deletion in exon 5 of the gene. Efforts should be devoted to unravel the mechanisms underlying the intrafamilial phenotypic variability observed in the families affected, and to identify new genes responsible for the still unsolved WS4 cases.


Assuntos
Mutação , Fatores de Transcrição SOXE/genética , Síndrome de Waardenburg/diagnóstico , Síndrome de Waardenburg/genética , Sequência de Aminoácidos , Substituição de Aminoácidos , Pré-Escolar , Análise Mutacional de DNA , Éxons , Ordem dos Genes , Doença de Hirschsprung , Humanos , Masculino , Dados de Sequência Molecular , Fenótipo , Espanha
16.
Orphanet J Rare Dis ; 8: 187, 2013 Dec 02.
Artigo em Inglês | MEDLINE | ID: mdl-24289864

RESUMO

Despite it has been reported that several loci are involved in Hirschsprung's disease, the molecular basis of the disease remains yet essentially unknown. The study of collective properties of modules of functionally-related genes provides an efficient and sensitive statistical framework that can overcome sample size limitations in the study of rare diseases. Here, we present the extension of a previous study of a Spanish series of HSCR trios to an international cohort of 162 HSCR trios to validate the generality of the underlying functional basis of the Hirschsprung's disease mechanisms previously found. The Pathway-Based Analysis (PBA) confirms a strong association of gene ontology (GO) modules related to signal transduction and its regulation, enteric nervous system (ENS) formation and other processes related to the disease. In addition, network analysis recovers sub-networks significantly associated to the disease, which contain genes related to the same functionalities, thus providing an independent validation of these findings. The functional profiles of association obtained for patients populations from different countries were compared to each other. While gene associations were different at each series, the main functional associations were identical in all the five populations. These observations would also explain the reported low reproducibility of associations of individual disease genes across populations.


Assuntos
Doença de Hirschsprung/metabolismo , Feminino , Predisposição Genética para Doença , Genótipo , Doença de Hirschsprung/genética , Humanos , Masculino , Polimorfismo de Nucleotídeo Único/genética
17.
Dev Biol ; 382(1): 320-9, 2013 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-23707863

RESUMO

Finding genes for complex diseases has been the goal of many genetic studies. Most of these studies have been successful by searching for genes and mutations in rare familial cases, by screening candidate genes and by performing genome wide association studies. However, only a small fraction of the total genetic risk for these complex genetic diseases can be explained by the identified mutations and associated genetic loci. In this review we focus on Hirschsprung disease (HSCR) as an example of a complex genetic disorder. We describe the genes identified in this congenital malformation and postulate that both common 'low penetrant' variants in combination with rare or private 'high penetrant' variants determine the risk on HSCR, and likely, on other complex diseases. We also discuss how new technological advances can be used to gain further insights in the genetic background of complex diseases. Finally, we outline a few steps to develop functional assays in order to determine the involvement of these variants in disease development.


Assuntos
Variação Genética , Doença de Hirschsprung/genética , Modelos Biológicos , Animais , Estudos de Associação Genética , Predisposição Genética para Doença , Doença de Hirschsprung/patologia , Humanos
18.
Biomed Res Int ; 2013: 585106, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23710452

RESUMO

Preimplantation genetic diagnosis (PGD) of single gene disorders, combined with HLA matching (PGD-HLA), has emerged as a tool for couples at risk of transmitting a genetic disease to select unaffected embryos of an HLA tissue type compatible with that of an existing affected child. Here, we present a novel one-step multiplex PCR to genotype a spectrum of STRs to simultaneously perform HLA typing and PGD for ß-thalassemia. This method is being routinely used for PGD-HLA cycles in our department, with a genotyping success rate of 100%. As an example, we present the first successful PGD-HLA typing in Spain, which resulted in the birth of a boy and subsequent successful HSC transplantation to his affected brother, who is doing well 4 years following transplantation. The advantage of our method is that it involves only a round of single PCR for multiple markers amplification (up to 10 markers within the HLA and 6 markers at the ß-globin loci). This strategy has allowed us to considerably reduce the optimization of the PCR method for each specific PGD-HLA family as well as the time to obtain molecular results in each cycle.


Assuntos
Teste de Histocompatibilidade/métodos , Reação em Cadeia da Polimerase Multiplex/métodos , Globinas beta/genética , Talassemia beta/diagnóstico , Adulto , Criança , Transferência Embrionária , Feminino , Fertilização in vitro , Humanos , Masculino , Oócitos/citologia , Oócitos/metabolismo , Gravidez , Diagnóstico Pré-Implantação , Técnicas de Reprodução Assistida , Espanha , Talassemia beta/sangue , Talassemia beta/genética
19.
Histol Histopathol ; 28(9): 1117-36, 2013 09.
Artigo em Inglês | MEDLINE | ID: mdl-23605783

RESUMO

Hirschsprung disease (HSCR), or aganglionic megacolon, is a developmental disorder characterised by the absence of ganglion cells along variable length of the distal gastrointestinal tract, leading to the most common form of functional intestinal obstruction in neonates and children. Aganglionosis is attributed to a failure of neural crest cells to migrate, proliferate, differentiate or survive during enteric nervous system (ENS) development in the embryonic stage. The incidence of HSCR is estimated at 1/5000 live births and most commonly presents sporadically with reduced penetrance and male predominance, although it can be familial and may be inherited as autosomal dominant or autosomal recessive. In 70% of cases, HSCR occurs as an isolated trait and in the other 30% HSCR is associated with other congenital malformation syndromes. HSCR has a complex genetic etiology with several genes and loci being described as associated with either isolated or syndromic forms. These genes encode for receptors, ligands (especially those participating in the RET and EDNRB signaling transduction pathways), transcriptional factors or other cell elements that are usually involved in the neural crest cell development and migration that give rise to ENS. Nevertheless, the RET proto-oncogene is considered the major disease causing gene in HSCR. A common RET variant within the conserved transcriptional enhancer sequence in intron 1 has been shown to be associated with a great proportion of sporadic cases and could act as a modifier by modulating the penetrance of mutations in other genes and possibly of those mutations in the RET proto-oncogene itself.


Assuntos
Doença de Hirschsprung/diagnóstico , Doença de Hirschsprung/genética , Animais , Diferenciação Celular , Proliferação de Células , Endotelinas/metabolismo , Feminino , Predisposição Genética para Doença , Variação Genética , Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Doença de Hirschsprung/metabolismo , Humanos , Íntrons , Ligantes , Masculino , Camundongos , Mutação , Fenótipo , Proto-Oncogene Mas , Proteínas Proto-Oncogênicas c-ret/genética , Proteínas Proto-Oncogênicas c-ret/metabolismo , Fatores Sexuais , Transdução de Sinais , Fatores de Transcrição/metabolismo , Fator de Crescimento do Endotélio Vascular Derivado de Glândula Endócrina/genética
20.
PLoS One ; 8(1): e54800, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23372769

RESUMO

Hirschsprung disease (HSCR, OMIM 142623) is a developmental disorder characterized by the absence of ganglion cells along variable lengths of the distal gastrointestinal tract, which results in tonic contraction of the aganglionic colon segment and functional intestinal obstruction. The RET proto-oncogene is the major gene associated to HSCR with differential contributions of its rare and common, coding and noncoding mutations to the multifactorial nature of this pathology. In addition, many other genes have been described to be associated with this pathology, including the semaphorins class III genes SEMA3A (7p12.1) and SEMA3D (7q21.11) through SNP array analyses and by next-generation sequencing technologies. Semaphorins are guidance cues for developing neurons implicated in the axonal projections and in the determination of the migratory pathway for neural-crest derived neural precursors during enteric nervous system development. In addition, it has been described that increased SEMA3A expression may be a risk factor for HSCR through the upregulation of the gene in the aganglionic smooth muscle layer of the colon in HSCR patients. Here we present the results of a comprehensive analysis of SEMA3A and SEMA3D in a series of 200 Spanish HSCR patients by the mutational screening of its coding sequence, which has led to find a number of potentially deleterious variants. RET mutations have been also detected in some of those patients carrying SEMAs variants. We have evaluated the A131T-SEMA3A, S598G-SEMA3A and E198K-SEMA3D mutations using colon tissue sections of these patients by immunohistochemistry. All mutants presented increased protein expression in smooth muscle layer of ganglionic segments. Moreover, A131T-SEMA3A also maintained higher protein levels in the aganglionic muscle layers. These findings strongly suggest that these mutants have a pathogenic effect on the disease. Furthermore, because of their coexistence with RET mutations, our data substantiate the additive genetic model proposed for this rare disorder and further support the association of SEMAs genes with HSCR.


Assuntos
Doença de Hirschsprung/genética , Mutação , Semaforina-3A/genética , População Branca/genética , Colo/metabolismo , Colo/patologia , Feminino , Doença de Hirschsprung/metabolismo , Humanos , Masculino , Proto-Oncogene Mas , Proteínas Proto-Oncogênicas c-ret/genética , Proteínas Proto-Oncogênicas c-ret/metabolismo , Semaforina-3A/metabolismo , Espanha
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...